malaya of university - university of malayastudentsrepo.um.edu.my/6707/1/eraj.pdf · ini...

173
DEVELOPMENT OF A SCAFFOLD INCORPORATING ZINC-OXIDE NANO-PARTICLES FOR CARTILAGE TISSUE ENGINEERING UNDER PHYSIOLOGICAL CONDITIONS ERAJ HUMAYUN MIRZA FACULTY OF ENGINEERING UNIVERSITY OF MALAYA KUALA LUMPUR 2016 University of Malaya

Upload: others

Post on 19-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

  • DEVELOPMENT OF A SCAFFOLD INCORPORATING

    ZINC-OXIDE NANO-PARTICLES FOR CARTILAGE TISSUE

    ENGINEERING UNDER PHYSIOLOGICAL CONDITIONS

    ERAJ HUMAYUN MIRZA

    FACULTY OF ENGINEERING

    UNIVERSITY OF MALAYA

    KUALA LUMPUR

    2016

    Unive

    rsity

    of Ma

    laya

  • DEVELOPMENT OF A SCAFFOLD INCORPORATING

    ZINC-OXIDE NANO-PARTICLES FOR CARTILAGE

    TISSUE ENGINEERING UNDER PHYSIOLOGICAL

    CONDITIONS

    ERAJ HUMAYUN MIRZA

    THESIS SUBMITTED IN FULFILMENT OF THE

    REQUIREMENTS FOR THE DEGREE OF DOCTOR OF

    PHILOSOPHY

    FACULTY OF ENGINEERING

    UNIVERSITY OF MALAYA

    KUALA LUMPUR

    2016

    Unive

    rsity

    of Ma

    laya

  • ii

    UNIVERSITY OF MALAYA

    ORIGINAL LITERARY WORK DECLARATION

    Name of Candidate: Eraj Humayun Mirza

    Matric No: KHA110058

    Name of Degree: Doctor of Philosophy

    Title of Thesis: The Development of a Scaffold incorporating Zinc-Oxide Nano-particles

    for Cartilage Tissue Engineering under Physiological Conditions

    Field of Study: Biomaterials and Tissue Engineering

    I do solemnly and sincerely declare that:

    (1) I am the sole author/writer of this Work;

    (2) This Work is original;

    (3) Any use of any work in which copyright exists was done by way of fair dealing and

    for permitted purposes and any excerpt or extract from, or reference to or reproduction of

    any copyright work has been disclosed expressly and sufficiently and the title of the Work

    and its authorship have been acknowledged in this Work;

    (4) I do not have any actual knowledge nor do I ought reasonably to know that the making

    of this work constitutes an infringement of any copyright work;

    (5) I hereby assign all and every rights in the copyright to this Work to the University of

    Malaya (“UM”), who henceforth shall be owner of the copyright in this Work and that

    any reproduction or use in any form or by any means whatsoever is prohibited without

    the written consent of UM having been first had and obtained;

    (6) I am fully aware that if in the course of making this Work I have infringed any

    copyright whether intentionally or otherwise, I may be subject to legal action or any

    other action as may be determined by UM.

    Candidate’s Signature Date: 22nd August 2016

    Subscribed and solemnly declared before,

    Witness’s Signature Date: 22nd August 2016

    Name:

    Designation:

    Unive

    rsity

    of Ma

    laya

  • iii

    Abstract

    Cartilage tissue is among the most complex of all in the human body. Degeneration of

    cartilage, lack of repair and various traumatic and pathological conditions among

    individuals increase joint pain and disability. Researchers have been trying to repair

    cartilage damage for decades, but have failed to achieve an optimal repair strategy.

    Cartilage tissue is adapted to low oxygen environments and this condition appears to be

    a key factor in the growth, regulation, and survival of chondrocytes; which are the only

    cells present in cartilage tissue. The present thesis defines the fabrication and

    characterisation of an antibacterial biomaterial for cartilage tissue repair. Furthermore,

    this thesis promotes the use of Zinc Oxide (ZnO) nanoparticles (NP) for better growth of

    cartilage cells and survival of cartilage tissue as a whole.

    Composite scaffolds and thin films of polyoctanediol citrate (POC) polyester elastomer,

    with varying concentrations (1wt%, 3wt% and 5wt%) of ZnO were fabricated by a

    solvent-casting/particulate-leaching and mould casting technique respectively. It was

    observed that material properties can be successfully controlled by simple variation of

    NP concentration within the composite. The ion release kinetics from ZnO-POC scaffolds

    are strongly dependent on NP concentration and degradation of pure POC matrix. All the

    composite scaffolds showed strong antibacterial characteristics. However, cell culture

    studies demonstrated that 1% ZnO incorporation in POC polymer is the optimal

    concentration for chondrocyte cells.

    Moreover, the effect of 1% ZnONP on chondrocyte proliferation and matrix synthesis

    cultured under normoxia (21% O2) and hypoxia (5% O2) demonstrated upregulation of

    chondrocyte proliferation and sulphated glycosaminoglycan (S-GAG) in hypoxic culture.

    A synergistic effect of oxygen concentration and 1% ZnONP in up-regulation of anabolic

    gene expression (Type II collagen (COL2A1) and aggrecan (ACAN)), and a down

    regulation of catabolic (MMP-13) gene expression was observed. Furthermore,

    Unive

    rsity

    of Ma

    laya

  • iv

    production of transcription factor hypoxia-inducible factor 1A (HIF-1A) in response to

    hypoxic condition to regulate chondrocyte survival under hypoxia was not affected by the

    presence of 1% ZnONP. Lastly this thesis discusses the physiological adaptations of

    cartilage tissue in a dynamic mechanical loading and hypoxic condition to yield benefits

    of combined bio-factors for cartilage tissue engineering applications. The results indicate

    that the combination of dynamic loading is compatible with the nanoparticle addition.

    Furthermore, dynamic loading suppresses MMP-13, and increase the expression of

    COL2A1 and ACAN with an increase in cell viability, and promotion of rounded cell

    morphology (a phenotypic marker).

    It was concluded that POC-ZnONP scaffolds are of major importance in the development

    of multifunctional scaffolds based on biodegradable polyesters for cartilage tissue

    engineering and presence of 1% ZnONP appears to preserve homeostasis of cartilage in

    its hypoxic environment. While 1% ZnONP should be considered for beneficial

    incorporation into 3D hypoxic culture systems in the presence of mechanical stimulation.

    Further studies must focus on determining the use of 1% ZnONP in different polymers

    for use in various tissue engineering applications.

    Unive

    rsity

    of Ma

    laya

  • v

    Abstrak

    Tisu rawan adalah antara tisu yang paling kompleks di dalam badan manusia.

    Kemerosotan rawan, kurang pembaikan dan pelbagai trauma dan keadaan patologi di

    kalangan individu meningkatkan kesakitan pada sendi dan kecacatan. Penyelidik telah

    berusaha untuk memperbaiki kerosakan rawan selama bertahun-tahun, tetapi telah gagal

    mencapai strategi optimum pembaikan. Tisu rawan disuaidirikan kepada persekitaran

    rendah oksigen dan keadaan ini menjadi satu faktor kepada pertumbuhan, peraturan dan

    kebertahanan kondrosit; iaitu satu-satunya jenis sel yang ada di dalam tisu rawan. Tesis

    ini menentukan fabrikasi dan pencirian biobahan antibakteria untuk pembaikan tisu

    rawan. Tambahan pula, tesis ini menggalakkan penggunaan nano-partikel (NP) Zink

    Oksida (ZnO) untuk pertumbuhan yang lebih baik bagi sel-sel rawan dan kebertahanan

    tisu rawan secara keseluruhannya.

    Perancah komposit dan filem nipis daripada elastomer polyester polyoctanediol sitrat

    (POC), dengan kepekatan (1wt%, 3wt% and 5wt%) ZnO yang berbeza telah

    difabrikasikan masing –masing dengan menggunakan teknik pelarut-pemutus/larutresap-

    zarahan dan pemutus acuan. Daripada pemerhatian, didapati sifat-sifat material telah

    berjaya terkawal oleh perubahan mudah kepekatan NP di dalam komposit itu. Kinetik

    pembebasan ion daripada perancah ZnO-POC bergantung secara kuat kepada kepekatan

    NP dan degradasi matriks tulen POC. Semua perancah komposit telah menunjukkan sifat-

    sifat antibakteria yang kuat. Walau bagaimanapun, kajian sel kultur mendapati gabungan

    1% ZnO di dalam polimer POC adalah kepekatan yang optimum untuk sel-sel kondrosit.

    Tambahan lagi, kesan 1% ZnONP terhadap pertumbuhan kondrosit dan sintesis matriks

    yang dikultur di bawah keadaan normoksia (21% O2) dan hipoksia (5% O2) telah

    menunjukkan pengawalan-atas pertumbuhan kondrosit dan sulfat-glycosaminoglycan (S-

    GAG) di dalam kultur hipoksia. Satu kesan sinergi kepekatan oksigen dan 1% ZnONP di

    dalam penyataan gen pengawalan-atas anabolic (Kolagen Type II (COL2A1) dan

    Unive

    rsity

    of Ma

    laya

  • vi

    aggrecan (ACAN)), dan penyataan gen pengawalan-bawah katabolic (MMP-13) telah

    diperhatikan. Tambahan pula, pengeluaran hipoksia transkripsi faktor-dorongan factor

    1A (HIF-1A) di dalam tindakbalasnya terhadap keadaan hipoksia untuk mengawal

    kewujudan kondrosit tidak dipengaruhi dengan kehadiran 1% ZnONP. Akhir sekali, tesis

    ini membincangkan tentang penyesuaian fisiologi tisu rawan di dalam pemuatan

    mekanikal dinamik dan keadaan hipoksia untuk menghasilkan manfaat biofaktor yang

    digabungkan untuk penggunaan kejuruteraan tisu rawan. Keputusan menunjukkan

    gabungan pemuatan dinamik adalah serasi dengan penambahan nano-partikel. Tambahan

    pula, pemuatan dinamik menyekat MMP-13 dan meningkatkan penyataan COL2A1 dan

    ACAN dengan peningkatan pertumbuhan sel dan morfologi sel bulat (satu penanda

    fenotip).

    Ini dapat disimpulkan bahawa perancah POC-ZnONP sangat penting di dalam

    penambahbaikan pelbagai fungsi perancah berasaskan polyester biodegradasi untuk

    kejuruteraan tisu rawan dan kehadiran 1% ZnONP telah memelihara homestasis rawan di

    dalam persekitaran hipoksia. Manakala 1% ZnONP perlu dipertimbangkan untuk

    gabungan bermanfaat kepada system 3D kultur hipoksia dengan kehadiran rangsangan

    mekanikal. Kajian lanjut fokus kepada penentuan penggunaan 1% ZnONP di dalam

    polimer yang berbeza untuk kegunaan pelbagai aplikasi kejuruteraan tisu.

    Unive

    rsity

    of Ma

    laya

  • vii

    I dedicate this thesis to Ammi and Baba

    Unive

    rsity

    of Ma

    laya

  • viii

    ACKNOWLEDGEMENT

    First of all I would like to praise ALLAH who blessed me with the strength to accomplish

    my dream.

    I would like to explicitly thank my supervisors Dr. Belinda Pingguan – Murphy and Dr.

    Wan Mohd Azhar bin Wan Ibrahim, for their unconditional support, vast knowledge,

    precious time and critical feedback. I am especially grateful to Dr. Belinda Pingguan–

    Murphy for her patience with me and her critical review which allowed me to enhance

    the quality of my articles and my thesis. Moreover, I sincerely acknowledge my ustaad,

    Dr. Ali Moradi, from whom I have learnt not only cell culture techniques and scaffold

    fabrication but also to help everyone. I would like to extend my appreciation to my

    friends, my lab mates and my colleagues; Adel Al-Halawani, Yasser Al-Saffar, Haris

    Akram, Salfarina Ezrina, Kar Wey, Poon Chi-Tat, Janu Ru, Dr. Chong Pan-Pan, Dr.

    Eleanor Parker, Syed Shahabuddin, Shahid Mehmood, Kashan Pirzada, Forough

    Ataollahi, Adel Dalilottojari and all those who supported me knowing or unknowingly.

    I like to thank Dr. Wan Safwani and Dr. Farina to provide me with their valuable

    suggestions during candidature defence.

    I admire the support from Dr. Adib, Dr. Ahmad Khairi, Liyana bint Abu, Kakak Enas

    naeem, Mr. Hanafi, Mr Adhli and all the staff at biomedical engineering administration.

    I would also like to thank all the laboratory technical staff throughout University of

    Malaya.

    I would like to acknowledge the scholarship from MOHE without which I would have

    never had enough financial support to continue my PhD. Furthermore, I would like to

    thank University of Malaya for providing IPPP and HIR grant to fulfil my research needs.

    I would like to thank Ammi and Baba for their unconditional love, support and long

    conversations to calm down my anxiety. A sincere thanks to my brothers. My gratitude

    and love to my wife who loves me and care for me.

    Unive

    rsity

    of Ma

    laya

  • ix

    TABLE OF CONTENTS

    LIST OF FIGURES ……………………………………………………… xv

    LIST OF TABLES ………………………………………………………. xx

    LIST OF ABBREVIATIONS …………………………………………… xxi

    CHAPTER 1: INTRODUCTION ………………………………………..

    1.1 Thesis Overview ………………………………………………………

    1.1 Background ……………………………………………………………

    1.2 Problem Statement …………………………………………………….

    1.3 Objectives ……………………………………………………………...

    1.4 Hypotheses …………………………………………………………….

    1

    1

    1

    3

    4

    5

    CHAPTER 2: LITERATURE REVIEW ……………………………….

    2.1 Introduction ……………………………………………………………

    2.2 Cartilage ……………………………………………………………….

    2.2.1 Composition of articular cartilage ………………………….

    2.2.1.1 Chondrocytes …………………………………………

    2.2.1.2 Extracellular Matrix ………………………………….

    2.2.1.3 Water …………………………………………………

    2.2.1.4 Collagens ……………………………………………..

    2.2.1.5 Proteoglycans ………………………………………...

    2.2.1.6 Non-Collagenous proteins and Glycoproteins ……….

    2.2.2 Zonal arrangement in articular cartilage ……………………….

    2.3 Chondrogenesis and chondrocyte differentiation ……………………...

    2.4 Articular Cartilage Disease and Injury ………………………………...

    2.5 Treatment Options for Cartilage Injury ………………………………..

    2.5.1 Marrow Stimulation Technique ………………………………..

    2.5.2 Osteochondral Grafting ………………………………………...

    6

    6

    6

    7

    7

    8

    8

    9

    9

    10

    10

    12

    12

    14

    14

    15

    Unive

    rsity

    of Ma

    laya

  • x

    2.5.3 Autologous Chondrocyte Transplantation ……………………..

    2.6 Cartilage Tissue Engineering ………………………………………….

    2.6.1 Cell Sources ……………………………………………………

    2.6.2 Cell Signalling Strategies ………………………………………

    2.6.2.1 Mechanical stimulus ………………………………….

    2.6.2.2 Oxygen Tension/Hypoxia ……………………………

    2.6.3 Tissue Engineering Scaffolds ………………………………….

    2.7 Poly Octanediol Citrate (POC) ………………………………………..

    2.7.1 POC, its structure, synthesis, and form ………………………...

    2.7.2 POC as a biomaterial …………………………………………..

    2.7.3 POC in cartilage tissue engineering ……………………………

    2.8 Infection and Anti-bacterial scaffolds …………………………………

    2.9 Antibacterial activity of ZnO ………………………………………….

    2.10 Motivation for current study …………………………………………..

    16

    17

    18

    19

    20

    22

    23

    25

    25

    26

    27

    28

    32

    33

    CHAPTER 3: MATERIALS AND METHODS ………………………...

    3.1 Introduction ……………………………………………………………

    3.2 Preparation of POC pre-polymer ……………………………………...

    3.3 Fabrication of POC and ZnO-POC composites ……………………….

    3.4 Chondrocyte Isolation …………………………………………………

    3.4.1 Chondrocyte Culture Medium …………………………………

    3.4.2 Cartilage ECM digestion ………………………………………

    3.4.2.1 Protease preparation ………………………………….

    3.4.2.2 Collagenous preparation ……………………………...

    3.4.3 Cell Viability …………………………………………………...

    3.5 Cell proliferation assay ………………………………………………..

    3.6 DNA Biochemical Assay ……………………………………………...

    36

    36

    36

    38

    40

    42

    42

    43

    43

    43

    44

    44

    Unive

    rsity

    of Ma

    laya

  • xi

    3.6.1 Lysis of BACs ………………………………………………….

    3.6.2 DNA quantification …………………………………………….

    3.6.2.1 DNA Standard ………………………………………..

    3.7 Sulphated Glycosaminoglycan (S-GAG) quantification assays ………

    3.8 Quantitative Polymerase Chain Reaction (q-PCR) ……………………

    3.8.1 RNA Isolation ………………………………………………….

    3.8.2 cDNA synthesis ………………………………………………..

    3.8.3 Gene Expression ……………………………………………….

    3.9 Scanning Electron Microscopy (SEM) for cell adhesion ……………...

    3.10 Live/Dead Assay ……………………………………………………..

    3.11 Statistical Analysis …………………………………………………

    45

    45

    46

    47

    48

    48

    49

    49

    50

    51

    51

    CHAPTER 4: CHARACTERISATION OF POLYOCTANEDIOL

    CITRATE-ZINC OXIDE NANO-COMPOSITE SCAFFOLD ………...

    4.1 Introduction …………………………………………………………....

    4.2 Materials and Methods ………………………………………………...

    4.2.1 Surface and elemental properties ………………………………

    4.2.2 Zinc oxide nanoparticles distribution within POC …………….

    4.2.3 Surface wettability test …………………………………………

    4.2.4 Mechanical property and porosity ……………………………..

    4.2.5 Swelling test ……………………………………………………

    4.2.6 Fourier transform infrared spectroscopy (FTIR) analysis ……..

    4.2.7 Thermal analysis ……………………………………………….

    4.2.8 Degradation analysis …………………………………………...

    4.2.9 ZnO NPs release kinetics in physiological conditions …………

    4.2.10 Anti-bacterial properties ……………………………………….

    4.2.11 In vitro tests with chondrocyte cell culture …………………….

    52

    52

    54

    54

    54

    55

    55

    56

    56

    56

    57

    57

    58

    58

    Unive

    rsity

    of Ma

    laya

  • xii

    4.3 Results …………………………………………………………………

    4.3.1 Surface morphology ……………………………………………

    4.3.2 Influence of ZnO-NPs on relative hydrophilicity ……………...

    4.3.3 Physical properties of scaffolds ………………………………..

    4.3.4 Swelling and cross-linking density …………………………….

    4.3.5 Chemical composition …………………………………………

    4.3.6 Thermal stability ……………………………………………….

    4.3.7 Degradation …………………………………………………….

    4.3.8 ZnO release kinetics ……………………………………………

    4.3.9 Anti-bacterial properties of ZnO-POC scaffolds ………………

    4.3.10 Cytotoxic effects of ZnO NPs on primary chondrocytes ………

    4.4 Discussion ……………………………………………………………..

    4.4.1 Surface morphology ……………………………………………

    4.4.2 Influence of ZnO-NPs on relative hydrophilicity ……………...

    4.4.3 Physical properties of ZnO-POC scaffolds: porosity and

    elasticity ……………………………………………………......

    4.4.4 Swelling and cross-linking density …………………………….

    4.4.5 Chemical composition …………………………………………

    4.4.6 Thermal stability ……………………………………………….

    4.4.7 Degradation …………………………………………………….

    4.4.8 ZnO release kinetics ……………………………………………

    4.4.9 Anti-bacterial properties of ZnO-POC composite scaffolds …...

    4.4.10 Cytotoxic effects of ZnO NPs on primary chondrocytes ………

    59

    59

    60

    62

    63

    63

    64

    65

    66

    68

    70

    71

    71

    71

    72

    73

    73

    74

    75

    75

    76

    77

    Unive

    rsity

    of Ma

    laya

  • xiii

    CHAPTER 5: EFFECT OF HYPOXIA ON BOVINE ARTICULAR

    CHONDROCYTES ……………………………………………………….

    5.1 Introduction ……………………………………………………………

    5.2 Materials and Methods ………………………………………………...

    5.2.1 Experimental Design …………………………………………...

    5.2.2 Cell Proliferation Assays ………………………………………

    5.2.3 Sulphated glycosaminoglycan (S-GAG) quantification

    assays …………………………………………………………..

    5.2.4 Quantitative Polymerase Chain Reaction (q-PCR) …………….

    5.2.5 Statistical Analysis ……………………………………………..

    5.3 Results …………………………………………………………………

    5.3.1 Cell proliferation ……………………………………………….

    5.3.2 Proteoglycan Synthesis ………………………………………...

    5.3.3 Gene expression ………………………………………………..

    5.4 Discussion …………………………………………………………......

    5.4.1 Cell proliferation ……………………………………………….

    5.4.2 Proteoglycan Synthesis ………………………………………...

    5.4.3 Gene expression ………………………………………………..

    79

    79

    80

    81

    81

    81

    81

    82

    82

    82

    83

    87

    88

    89

    90

    91

    91

    CHAPTER 6: EFFECT OF DYNAMIC MECHANICAL

    COMPRESSION ON BOVINE ARTICULAR CHONDROCYTES ….

    6.1 Introduction ……………………………………………………………

    6.2 Materials and Methods ………………………………………………...

    6.2.1 Experimental Design …………………………………………...

    6.2.2 Application of dynamic compression ………………………….

    6.2.3 Cell Proliferation Assay ………………………………………..

    95

    95

    97

    97

    98

    99

    Unive

    rsity

    of Ma

    laya

  • xiv

    6.2.4 Sulphated glycosaminoglycan (S-GAG) quantification

    assays ……………………………………………………………...

    6.2.5 Quantitative Polymerase Chain Reaction (q-PCR) …………….

    6.3 Results …………………………………………………………………

    6.3.1 Cell proliferation and morphology …………………………….

    6.3.2 Proteoglycan synthesis …………………………………………

    6.3.3 Gene Expression ……………………………………………….

    6.4 Discussion ……………………………………………………………..

    6.4.1 Cell proliferation and morphology …………………………….

    6.4.2 Proteoglycan synthesis …………………………………………

    6.4.3 Gene Expression ……………………………………………….

    100

    100

    100

    100

    105

    106

    107

    108

    109

    109

    CHAPTER 7: CONCLUSIONS AND FUTURE DIRECTIONS ………

    7.1 Conclusions ………………………………………………………........

    7.2 Future directions ……………………………………………………….

    111

    111

    115

    REFERENCES …………………………………...………………………. 116

    PUBLICATIONS ……………………...…………………………………..

    APPENDIX I ………………………………………………………………

    APPENDIX II ……………………………………………………………...

    149

    153

    154

    Unive

    rsity

    of Ma

    laya

  • xv

    LIST OF FIGURES

    Figure 2.1: Zones of Articular cartilage, modified from (Bhosale & Richardson,

    2008…………...………………………………………………………..........................11

    Figure 2.2: Wutizite structure of ZnO crystal ………………………………………….32

    Figure 2.3: Chondrocytes cultured in a T-75 flask losing their morphology and turning

    flat only after 24 hours of culture ………………………………………………………34

    Figure 3.1: (A) The reaction scheme of POC pre-polymer fabrication via the

    polycondensation reaction in a two necked round bottom flask with a nitrogen purge to

    remove excess water content. (B) Synthesis scheme of monomers reacting to form POC

    polymer ……………………………………………………..………………………….37

    Figure 3.2: Steps involved in fabrication of (A) Pure-POC and (B) POC-ZnONP

    scaffolds, films and coatings prior to curing ……………………………....……………39

    Figure 3.3: A custom-designed rack placed under water to categorise Pure-POC scaffolds

    from ZnO-POC scaffolds and to leach out the salt from scaffolds ……….......…………40

    Figure 3.4: Procedure of isolation of primary chondrocytes from cow legs. (A) Removal

    of hide from cow leg insert shows completely removed hide, (B) Opening of joint by

    removing ligament, (C) Cutting of cartilage and (D) Complete shaved cartilage joint and

    explants placed in sterile container ……………………………………………......……41

    Figure 4.1: FESEM images of POC (control) and 1%, 3% and 5% ZnO-POC composite

    scaffolds; (A) POC, (B) 1% ZnO-POC, (C) 3% ZnO-POC and (D) 5% ZnO-POC; inserts

    show representative magnified surfaces of pore walls within scaffolds (inserts: A and B

    bars = 1 µm; C and D bars = 2 µm) ……………………………………………...………60

    Unive

    rsity

    of Ma

    laya

  • xvi

    Figure 4.2: Optical microscopy images of Pure-POC and ZnO-POC surfaces developed

    by spin-coating technique at a magnification of 100X; (A) POC, (B) 1% ZnO-POC, (C)

    3% ZnO-POC and (D) 5% ZnO-POC …………………………………………..………61

    Figure 4.3: Water-in-air contact angle results measured for thin films of Pure-POC and

    1%, 3% and 5% ZnO-POC produced by spin-coating technique ……………………….62

    Figure 4.4: Percentage swelling in water for Pure-POC (control) and 1%, 3% and 5%

    ZnO-POC composite films ……………………………………………………………..63

    Figure 4.5: Representative ATR-FTIR spectra of POC and 5% ZnO-POC scaffolds …64

    Figure 4.6: Thermal degradation curves of Pure-POC and (1%, 3% & 5%) ZnO-POC

    scaffolds measured by TGA ……………………………………………………………65

    Figure 4.7: Weight loss of Pure-POC, 1%, 3% and 5% ZnO-POC scaffolds over the

    period of 20 weeks in Phosphate Buffered Saline (PBS) ………………………………66

    Figure 4.8: In-vitro Release kinetics profile of ZnO (Zn2+) from ZnO-POC scaffolds in

    PBS at 37°C; Mt = mass of ZnO released at time intervals; M∞ = total mass of ZnO within

    the scaffold (all Mt /M∞ ratios are the mean values for five samples measured with

    AAS)………………………………………………………........………………………67

    Figure 4.9: Zn-ions released over the period of 28 days calculated in ppm via AAS.

    Where * shows a significant difference between the number of days, p

  • xvii

    Figure 4.11: (A) Resazurin reduction (%) for Pure-POC (control) and 1%, 3% and 5%

    ZnO-POC scaffolds after 24 and after 72 hours. FESEM images of scaffolds after 72

    hours in chondrocyte culture: (B) Pure-POC; and (C) 1% ZnO-POC (bar = 10

    µm)……………………………………………………………………….…………….70

    Figure 5.1: Summary of experimental design ……………………………...…………81

    Figure 5.2: Cell vitality determined via resazurin reduction assay, of chondrocytes,

    seeded on Pure-POC and 1% ZnO-POC in normoxic (21%O2) & hypoxic (5%O2)

    conditions. Where * shows a significant difference between O2 tension. 5≤ N ≤ 6,

    p

  • xviii

    differences between groups and * shows a significant difference between O2 tension. 5≤

    N ≤ 6, p

  • xix

    Figure 6.5: Confocal imaging of Chondrocytes (A-H) seeded on Pure-POC and 1% ZnO-

    POC scaffolds in normoxic (21%O2) & hypoxic (5%O2) conditions, under static and

    dynamic culture regimes. Outer images are at 10X with a scale bar of 300 μm and their

    respective inserts are at 40X magnification with a scale bar of 100 μm……………….102

    Figure 6.6: SEM imaging of Chondrocytes (A-H) seeded on Pure-POC and 1% ZnO-

    POC scaffolds in normoxic (21%O2) & hypoxic (5%O2) conditions, under static and

    dynamic culture regimes. Outer images are taken at 1000X magnification, it has a scale

    bar of 10 μm. Corresponding images in inserts were taken at 3000X magnification having

    a scale bar of 2 μm……………………………………………………………………104

    Figure 6.7: DMMB assay for glycosaminoglycans (A) Total S-GAG, measured in

    digested scaffolds and (B) S-GAG measured in medium via DMMB assay for

    chondrocytes seeded on pure-POC and 1% ZnO-POC in normoxic (21%O2) & hypoxic

    (5%O2) conditions, under static and dynamic culture regimes. Where ^ shows a

    significant difference between due to the presence of ZnONP, * shows a significant

    difference between O2 tension and + shows a significant difference between static and

    dynamic culture. p

  • xx

    LIST OF TABLES

    Table 2.1: Constituents of articular cartilage with their respective composition……….8

    Table 3.1: Ingredients to prepare chondrocyte culture medium…………….…………..42

    Table 3.2: Ingredients to prepare papain digest buffer………………….………………45

    Table 3.3: Ingredients used in preparation of DMMB solution for DMMB

    assay………………………………………………………………….....………………47

    Table 3.4: Gene identities and their respective amplicon length from

    LifeTechnologies…………………………………….....………………………………50

    Table 4.1 Compression properties and porosities of ZnO-POC scaffolds……………....62

    Unive

    rsity

    of Ma

    laya

  • xxi

    List of Abbreviations

    3D three dimensional

    AAS atomic emission spectroscopy

    ACAN Aggrecan gene

    ACI Autologous Chondrocyte Implantation

    ANOVA Analysis of Variance

    ATCC American Type Culture Collection

    BAC Bovine Articular Chondrocytes

    BMPs Bone Morphogenic Proteins

    CA Citric Acid

    cDNA complimentary Deoxyribonucleic acid

    CLSM confocal laser scanning microscopy

    COL2A1 Collagen Type II – Alpha 1 gene

    COLX Collagen Type X gene

    COMP Cartilage Oligomeric Protein

    DMEM Dulbecco’s Modified Eagle’s Medium

    DMMB Dimethyl Methylene Blue

    DNA Deoxyribonucleic acid

    ECM Extra Cellular Matrix

    EDTA Ethylenediaminetetraacetic acid

    EDX Energy Dispersive X-ray

    em emission

    EPS equilibrium percentage swelling

    ex excitation

    FBS Foetal bovine serum

    FDA Food and Drug Administration

    Unive

    rsity

    of Ma

    laya

  • xxii

    FESEM Field Emission Scanning Electron microscope

    FTIR Fourier transform infrared spectroscopy

    GAGs Glycosaminoglycans

    GAPDH Glyceraldehyde-3-phosphate dehydrogenase gene

    GRAS Generally Regarded as Safe

    HA Hydroxyapatite

    HCl Hydrochloride

    HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

    HIF-1A Hypoxia Inducible Factor-1 alpha gene

    HNMR Hydrogen nuclear magnetic resonance

    LB-Broth Luria-Bertani broth

    MALDI-TOF Matrix-assisted laser desorption ionization- Time of Flight

    min minutes

    MMP-13 Matrix Metalloproteinases – 13 gene

    MP-AES microwave plasma-atomic emission spectrophotometer

    N2 Nitrogen

    NaCl Sodium Chloride

    NaOH Sodium Hydroxide

    NP Nanoparticles

    O2 Oxygen

    OD 1, 8 Octanediol

    PBS Phosphate Buffered Saline

    PCL Polycaprolactone

    PEG Poly Ethylene Glycol

    PGA Poly Glycolic Acid

    PGS Poly (glycerol sebacate)

    Unive

    rsity

    of Ma

    laya

  • xxiii

    PLA Poly D, L – Lactic acid

    PLGA Poly D, L – Lactic acid-co-glycolic acid

    PLLA Poly-L- Lactic acid

    POC Poly 1, 8 Octanediol Citrate

    PPLG Poly γ-propargyl-l-glutamate

    Ppm parts per million

    PVP polyvinylpyrolidone

    q-PCR Quantitative Polymerase Chain Reaction

    RA Rheumatoid Arthritis

    RNA ribonucleic acid

    ROS reactive oxygen species

    SEM Scanning Electron Microscopy

    S-GAG Sulphated Glycosaminoglycan

    SOX9 SRY-box 9

    SRTR Scientific Registry of Transplant Recipients

    SSC Saline Sodium Citrate

    TCP Tricalcium Phosphate

    TE Tissue Engineeirng

    TGA thermo gravimetric analysis

    UNOS United Network for Organ Sharing

    w/v weight by volume ratio

    w/w weight by weight ratio

    WACA Water-in-air contact angle

    ZnO Zinc Oxide

    ZnONP Zinc Oxide Nanoparticles

    β-TCP Beta Tricalcium Phosphate

    Unive

    rsity

    of Ma

    laya

  • 1

    CHAPTER 1: INTRODUCTION

    1.1 Thesis Overview

    This thesis comprises 7 chapters. Chapter 1 describes the background of the research and

    the research problem, followed by objectives and hypothesis. The second chapter

    discusses the related literature in depth, and lastly, the motivation to perform the research

    is included. A complete methodology and list of materials used is provided in chapter 3

    so the reader can repeat the experiment if they want to. Finally, chapter 4, 5 and 6 are

    divided according to the objectives, and each chapter includes the results and discussion

    for the defined objectives. Chapter 7 is the last chapter in this thesis and provides the

    essence of the thesis in the form of a conclusion, and future direction for related research.

    1.2 Background

    As per 17 May 2012, data provided by Organ Procurement and Transplantation

    Network/Scientific Registry of Transplant Recipients (SRTR)/ glyco (UNOS) shows,

    there are more than 114,000 people in the queue for organ transplants. Every 10 minutes

    a new patient is added to the queue and 18 people die each day due to organ shortages

    (Network, 2012). Each year, more than one million people are in need of heart valves,

    corneas, skin cardiovascular tissue, and bone tissue (Network, 2012).

    The facts and figures mentioned above only represent the United States of America. The

    rest of the world population, especially the third world countries where quality of life is

    poor and organ donations are scarce, are also affected. There is an urgent need for

    biocompatible, efficacious, and cost-effective engineered tissues and organs, and this

    need only increases with advances in quality of life and life expectancy.

    Articular cartilage is by far one of the most essential and yet functionally complex tissues

    that scientists are trying to repair, yet minimal success has been for decades. A major

    cause of disability is the joint pain that cartilage disease produces, which is not only

    Unive

    rsity

    of Ma

    laya

  • 2

    limited to older people but found in middle aged persons as well. The cause of joint pain

    is degeneration of articular cartilage due to primary osteoarthritis or trauma (Temenoff &

    Mikos, 2000). Cartilage is a tissue with limited self-repair capabilities, and hence any

    kind of injury to the cartilage tissue which is sustained for a substantial time eventually

    leads to further detrimental effects (O'Driscoll, 1998; Steinert et al., 2007; Temenoff &

    Mikos, 2000). Different approaches are adopted by clinicians to address the issue of

    cartilage repair depending upon the nature of cartilage damage. However current

    treatment options fail to secure a permanent solution. Furthermore, infection and

    inflammation may lead to treatment failing. One of the major causes of biomaterial

    infections is peri-operative contamination and research has shown that tissue engineered

    implants also pose a risk similar to biomaterial associated infections (Kuijer et al., 2007).

    Amongst total knee arthroplasty patients, a risk of peri-operative infection of up to 38%

    has been reported recently (Janz et al., 2015). Matrix-induced Autologous Chondrocyte

    Implantation has also been reported to cause superficial infection among patients (Bartlett

    et al., 2005). Additionally, a recent study demonstrated that only the presence of nano

    surface topography can help reduce bacterial adhesion while increasing cell proliferation

    due to nanoscale roughness (Liu et al., 2015). This finding adds to the use of

    nanomaterials, which are already employed in the delivery of drugs (Pi et al., 2015; Shi

    et al., 2015), fighting of infection (Kalashnikova et al., 2015), enhancing mechanical

    properties of scaffolds (Pooyan et al., 2015) and increasing cell proliferation (Holmes et

    al., 2016).

    Chondrocytes are the only cell type that is present in cartilage. These cells flourish in a

    hypoxic environment, while the Extra Cellular Matrix (ECM) in which these cells are

    embedded undergoes dynamic compression. Current research lack these native

    physiologic stimuli and other biofactors as required for chondrocytes to flourish.

    Unive

    rsity

    of Ma

    laya

  • 3

    Tissue engineering provides an alternative approach to address existing problems in an

    effective and efficient way. Even though cartilage tissue engineering have been adopted

    by scientist to cure the dilemma of cartilage damage. However, till date there are many

    problems that are associated directly and indirectly that impede implementation of

    cartilage repair strategy via tissue engineering approach.

    1.3 Problem Statement

    Various solutions have been put forward by scientists to successfully implement cartilage

    tissue engineering clinically. However, the limitations of their proposed techniques

    exceed their benefits. the use of hydrogels, natural polymers, synthetic polymers, and

    metal implants has been suggested but they have generally proved unsuitable for clinical

    use, due to their inferior mechanical properties (Johnstone et al., 2013; Spiller et al., 2011)

    and lack of recovery of the implanted material to its original level due to creep effect (Shi

    et al., 2016). Moreover, current cartilage repair strategies have raised a multitude of

    concerns, including infection (Lichstein et al., 2015; Yeo et al., 2015) implant failure

    (Hazelwood et al., 2015; Vahdati & Wagner, 2013), lack of native tissue architecture

    (Chung & Burdick, 2008; Doran, 2015) and absent cell morphology (Hardingham et al.,

    2002). In order to fight any bacterial infection many antibacterial agents have been

    studied (Aksoy et al., 2010; Davies, 2003). However, Zinc Oxide stands out as favourable

    because it also plays a significant role in cartilage synthesis(Kirsch et al., 2000). Current

    clinical methods include minimally invasive surgery to resurface articular cartilage,

    arthoscopic lavage, or marrow stimulation techniques. Unfortunately all the

    aforementioned clinical methods currently used are palliative and for temporary relief

    only. The most frequently used treatment options for cartilage tissue repair have

    limitations that hinder their long-term clinical implementation (Marlovits et al., 2006;

    Portocarrero et al., 2013).

    Unive

    rsity

    of Ma

    laya

  • 4

    Furthermore, only a handful of studies are present that implement all three components

    of cartilage tissue engineering: (a) cells, (b) scaffold, and (c) bio factors, together. More

    recent findings signify the importance of using a three-dimensional environment with

    dynamic loading for cartilage tissue engineering (Panadero et al., 2016).

    A cartilage tissue engineering approach must benefit from a suitable biocompatible and

    biodegradable scaffold that will provide a micro structure for the chondrocytes to settle

    and proliferate, while being porous enough to allow cell-cell interaction and exchange of

    nutrients for uniform tissue formation. Moreover, the newly fabricated scaffold must also

    be flexible enough to be implanted via minimally invasive techniques, where required

    (Johnstone et al., 2013).

    A successful intervention for a cartilage repair strategy must involve external stimuli that

    will deliver a physiologic environment to cultured chondrocytes under a 3D structure.

    Furthermore, to tackle inflammation and infection, a suitable anti-inflammatory and

    antibacterial mediator must be integrated to assist with diminishing these effects while

    having no adverse effects on the cells.

    In essence, the combination of biocompatible scaffold, physiologic stimuli and

    facilitation of antibacterial and anti-inflammatory properties bears promise for more

    effective treatment of cartilage disease.

    1.4 Objectives

    The objectives of this research are:

    1. To develop a biodegradable scaffold with controlled pore size and porosity

    mimicking the properties of native articular cartilage.

    2. To select and optimise the concentration of nano particles that must be

    biocompatible and having antibacterial effect while showing no toxicity towards

    chondrocytes.

    Unive

    rsity

    of Ma

    laya

  • 5

    3. To study the independent and combined effect of Hypoxia and ZnONP on bovine

    articular chondrocytes seeded on polymer scaffolds.

    4. To establish a relationship between dynamic mechanical stimuli, ZnONP and

    oxygen tension in chondrocyte seeded polymer scaffolds.

    1.5 Hypotheses

    1. It is hypothesised that chondrocytes will proliferate, secrete more extracellular

    matrix, and show higher gene expression under a hypoxic environment.

    Furthermore the incorporation of nanoparticles will deliver an anti-bacterial

    mechanism to the fabricated scaffold.

    2. Furthermore it is hypothesized that scaffolds that will go under dynamic

    compression with a combination of hypoxia and nanoparticles, will eventually

    display a more chondrogenic morphology and will help in elevating cartilage

    specific matrix gene while lowering catabolic gene expression.

    Unive

    rsity

    of Ma

    laya

  • 6

    CHAPTER 2: LITERATURE REVIEW

    2.1 Introduction

    In the present study, a novel composite biomaterial for use within cartilage tissue

    engineering is developed, optimised, and evaluated. The literature which underpins this

    is therefore reviewed, starting with the current understanding of cartilage as a tissue,

    followed by cartilage injury, and a review of cartilage repair options. From here, the focus

    moves to the three components of a tissue engineering strategy as applied to cartilage

    tissue engineering; first, cell source; second, cell signalling strategy (covering

    biochemical and biomechanical signalling); and finally, the scaffold material (including

    a focus on the target material for the present study).

    2.2 Cartilage

    Cartilage is a tissue that lacks blood vessels, nerves and lymph vessels, and is composed

    of sparingly distributed chondrocytes (only 1% in humans) (Poole, 1997). There are 3

    types of cartilage in the human body, depending upon matrix composition; elastic

    cartilage, fibrocartilage and hyaline cartilage (Jung, 2014). Elastic cartilage makes up the

    flexible cartilaginous structures of the nose and ear. It contains elastin as an additional

    component to ECM. Fibrocartilage contains a higher content of collagen in ECM than in

    hyaline cartilage and is present in ligaments and tendons that are located in close

    proximity to bone (Temenoff & Mikos, 2000). Hyaline cartilage is mainly found in the

    larynx, trachea, ribs, knee and bronchus (Jung, 2014).

    Long bones have a covering at their articulating ends known as hyaline cartilage. This

    type of cartilage is more commonly referred to as articular cartilage. This hyaline cartilage

    provides a low-friction surface with wear resistant properties (Bhosale & Richardson,

    2008).

    Unive

    rsity

    of Ma

    laya

  • 7

    Hyaline cartilage is made up of water and ECM. Within this ECM, type II collagen is

    most abundant, followed by proteoglycans that suffice for mechanical properties in vivo

    (Bhosale & Richardson, 2008). For an articular joint to function optimally, it is critical

    that engineered cartilage must comply with the properties of native articular cartilage. It

    must qualify to withstand load-bearing forces, have low friction a coefficient, and have

    other necessary biological and mechanical properties (Doran, 2015).

    2.2.1 Composition of articular cartilage

    2.2.1.1 Chondrocytes

    Among all the tissues of the human body, articular cartilage has the lowest cellular

    density, according to volume. Humans have only 1% cellular material in their articular

    cartilage, namely cells called chondrocytes (Temenoff & Mikos, 2000). Chondrocytes are

    sparse but extremely important for replacement of degraded matrix molecules in order to

    maintain homeostasis. At week 5 of gestational, a blastema is formed when few

    mesenchyme cells form an aggregate. This blastema starts secreting a cartilage matrix

    which forms the foundation of chondrocytes (Bhosale & Richardson, 2008). It is reported

    that a cilia extends from surface of some chondrocytes into the ECM that contributes to

    modifying ECM properties in response to mechanical stimulus (Buckwalter & Mankin,

    1997). Chondrocytes pass through various lineages, producing proteins vital for ECM.

    Chondrocytes in the periphery secrete collagen to form a hyaline cartilage. When

    chondrocytes mature, they tend to halt their activity, appear circular and are enclosed in

    a matrix (Buckwalter & Mankin, 1997; Hall, 1994). Table 2.1 summarises the

    constituents of articular cartilage.

    Unive

    rsity

    of Ma

    laya

  • 8

    Table 2.1: Constituents of articular cartilage with their respective composition

    (Jung, 2014; Poole, 1997; Temenoff & Mikos, 2000).

    Constituent Wet weight (%)

    Dry weight

    (%)

    Cellular

    Material Chondrocytes ----- 1

    Extracellular

    Matrix Water 80 -----

    Macromolecules Collagen Proteoglycans Non-collagenous

    proteins

    20-40 10-20 10-20 -----

    ----- 50-60 25-35 15-20

    2.2.1.2 Extracellular Matrix

    Apart from chondrocytes, articular cartilage comprises a complex mix of water and

    structural macromolecules in a framework that provides stability to the articular cartilage.

    A combination of water and macromolecules is called ECM. The majority constituent in

    ECM of articular cartilage is water, approximately 80% of the weight. However in

    different regions of cartilage the percentage of water varies from 60% to 80%. The

    macromolecules comprise collagens, proteoglycans, and non-collagenous proteins.

    Macromolecules (when combined) account for 20 – 40 % wet weight of articular cartilage

    (Jung, 2014).

    2.2.1.3 Water

    In articular cartilage water accounts for majority of the wet weight of the tissue; around

    80%. Apart from water tissue fluid contains bulk of cations to neutralise negatively

    charged GAG’s in ECM. Tissue fluid also contains metabolites and gases. The presence

    of tissue fluid is of vital importance to avascular cartilage as it helps the exchange of

    nutrients and oxygen (O2) with synovial fluid. Furthermore, recovery of cartilage and

    resistance to compression is achieved via the presence of tissue fluid in ECM (Buckwalter

    & Mankin, 1998b).

    Unive

    rsity

    of Ma

    laya

  • 9

    2.2.1.4 Collagens

    The second-most abundant constituent of ECM is collagen, which accounts for 10% to

    20% of the wet weight of articular cartilage (Bhosale & Richardson, 2008) while it

    equates to 50% to 60% of the dry weight of articular cartilage (Jung, 2014). Collagen

    types II, VI, IX, X and XI are found in articular cartilage. However, the major role player

    is type II collagen, which represents 90 – 95% of all the collagens present. Collagen type

    II also forms the foundation of interlinked fibrils in articular cartilage (Buckwalter &

    Mankin, 1997). The interlinked fibrils formed by type II collagen not only provide

    physical strength to the cartilage but also trap other macromolecules (Cohen et al., 1998;

    Temenoff & Mikos, 2000).

    2.2.1.5 Proteoglycans

    The third constituent of ECM are the proteoglycans, proteins that give compressive

    capabilities to the cartilage. However they are only 10 – 20 % of the total wet weight

    (Bhosale & Richardson, 2008), and they account for 25% to 35% in dry form (Buckwalter

    & Mankin, 1997). The proteoglycans are composed of polysaccharides and proteins in

    the ratio of 19 to 1 respectively (Wirth & Rudert, 1996). The protein forms the core, with

    attachment to one or more glycosaminoglycans (GAGs) chains (Buckwalter & Mankin,

    1997; Temenoff & Mikos, 2000). GAGs are from a family of polysaccharides which have

    a repeating disaccharide unit (Afratis et al., 2012; Anower-E-Khuda & Kimata, 2015).

    Chondroitin sulfate, dermatan sulfate, keratin sulfate and hyaluronic acid are all GAGs

    that are present in articular cartilage (Brézillon et al., 2014; Hall, 2012). Proteoglycans

    are sub-divided into 2 groups: (a) large aggregating proteoglycans and (b) small

    aggregating proteoglycans. Large aggregating proteoglycans are more commonly

    referred as Aggrecan (Buckwalter & Mankin, 1997). The Aggrecans are responsible for

    resilience and distribution of stress in articular cartilage (Temenoff & Mikos, 2000).

    Unive

    rsity

    of Ma

    laya

  • 10

    2.2.1.6 Non-Collagenous proteins and Glycoproteins

    Non-Collagenous proteins and Glycoproteins account for about 15% to 20% of articular

    cartilage in dry weight. In an articular cartilage there exists wide variety of these

    molecules. They possess minute quantities of oligosaccharide attached to a protein core.

    Contrary to proteoglycans and collagens, these non-collagenous proteins and

    glycoproteins have not been studied extensively. However, they tend to help in

    maintenance and organization of macromolecular network of ECM(Buckwalter et al.,

    2005; Gallagher et al., 1983). Among them, cartilage oligomeric protein (COMP) is most

    abundant. It is concentrated in the matrix of chondrocyte cells and has a binding affinity

    chondrocytes (Buckwalter & Mankin, 1997; Temenoff & Mikos, 2000); COMP is

    involved in maintaining the chondrogenic phenotype (Aigner & Fan, 2003).

    2.2.2 Zonal arrangement in articular cartilage

    Articular cartilage is divided in to 4 zones (figure 2.1) according to their structure and

    function.

    1. Superficial zone

    2. Transitional zone

    3. Deep zone

    4. Calcified zone

    1. Superficial zone

    The superficial zone comprises of flat cells and is the thinnest layer. It is laminated by a

    specialised layer of synovial fluid called the lamina splendens (Instructional Course

    Lectures, The American Academy of Orthopaedic Surgeons - Articular Cartilage. Part I:

    Tissue Design and Chondrocyte-Matrix Interactions*†, 1997). The lamina splendens is a

    protein that enhances the low – friction properties of the surface of articular cartilage

    (Buckwalter & Mankin, 1997; Temenoff & Mikos, 2000).

    Unive

    rsity

    of Ma

    laya

  • 11

    2. Transitional zone

    This zone is dominated by spheroid-shaped cells, although the cell density is lower.

    Collagen fibres with a higher diameter can be found in this zone, which also has a high

    content of the proteoglycan aggrecan (Bhosale & Richardson, 2008; Buckwalter &

    Mankin, 1997).

    3. Deep zone

    This zone has the lowest of cells densities, with spherical cells distributed throughout the

    zone. The proteoglycan concentration is the highest in this region (Bhosale & Richardson,

    2008; Buckwalter & Mankin, 1997).

    4. Calcified zone

    Mineralisation is higher in this region of articular cartilage, meaning that cells are

    embedded in a matrix. The cells in calcified zone synthesise Type X collagen, which is

    mainly responsible for providing mechanical stability to cartilage, and also assists in

    absorbing shocks(Buckwalter & Mankin, 1997; Temenoff & Mikos, 2000).

    Figure 2.1: Zones of Articular cartilage, modified from (Bhosale & Richardson,

    2008).

    Unive

    rsity

    of Ma

    laya

  • 12

    2.3 Chondrogenesis and chondrocyte differentiation

    The process by which cartilage forms is called chondrogenesis. This is a natural process

    and chondroprogenitor cells are responsible for its initiation. Chondroprogneitor cells are

    basically mesenchymal cells. Mesenchymal cells come together and form an aggregate

    and start condensing in one area; this marks the beginning of chondrogenesis. Bone

    Morphogenic Proteins (BMPs) are responsible for contributing to initiating

    chondrogenisis by helping in aggregation of loose mesenchymal cells. In the early stages

    of chondrogenesis, the mesenchymal cells secrete ECM and cell adhesion molecules

    (Goldring et al., 2006). Among the secreted ECM there are large amounts of type II

    collagen (Ng et al., 1993), N-cadherin (Oberlender & Tuan, 1994), N-cam (Widelitz et

    al., 1993) and tenascin (Mackie et al., 1987). Apart from the ECM and cell adhesion

    molecules, SRY-box 9 (SOX9); a transcription factor, is highly expressed. SOX9 plays a

    vital role in chondrogenesis and differentiation of chondrocytes.

    Differentiation of mesenchymal cells into chondrocytes causes the cells to secrete the

    ECM that contains large quantities of type II collagen and Aggrecan. After early

    chondrocyte differentiation, this is the next development stage of cartilage. In this stage

    the cells proliferate rapidly providing a template for the cartilage. After a certain time old

    cells draw away from the cell proliferation cycle and enter in to a phase of hypertrophic

    differentiation. In this process the chondrocytes grow in size, terminally differentiate,

    mineralize, and eventually undergo apoptosis (Freed et al., 1998; Zuscik et al., 2008).

    After the death of chondrocytes, they degrade and leave a matrix of their cells that serves

    as a scaffold for deposition of minerals and formation of bone (Zuscik et al., 2008).

    2.4 Articular Cartilage Disease and Injury

    Articular cartilage diseases represent the most common cause of joint pain in middle aged

    and older people (Buckwalter & Mankin, 1998b), and hospital treatment cost for total

    Unive

    rsity

    of Ma

    laya

  • 13

    knee replacement was reported to be US$ 28.5 billion in 2009 in the US alone (Murphy

    & Helmick, 2012). Together with acute injury, such cartilage disorders also result in a

    substantial reduction in quality of life (Minas, 1999; Murray et al., 2015).

    Depending upon the nature of injury, cartilage injury can be subdivided in to three major

    categories; Full thickness defect, partial defect and matrix disruption. A full thickness

    defect is the most painful and it usually affects entire cartilage thickness while also

    penetrating deep into the subchondral bone (Temenoff & Mikos, 2000). In this type of

    injury fibrin clot formation occurs at the site of defect. Blood and bone marrow cells bind

    to this site and form patches of fibrin clots (Aigner & Fan, 2003). The newly formed

    tissue lacks the strength of native articular cartilage and has a higher permeability. Lack

    of strength and excess permeability eventually contribute to degradation and permanent

    damage to articular cartilage and subchondral bone. An injury of as little as 3mm depth

    is considered to be a full thickness defect. (Coutts et al., 1997; Fortier et al., 2011;

    Temenoff & Mikos, 2000).

    A secondary injury is a partial thickness defect, resulting in disruption to the surface of

    cartilage. This damage, unlike a full thickness defect, does not extend to the subchondral

    bone. In this type of injury the surrounding cells do proliferate but not to an extent which

    can repair the defect completely (Temenoff & Mikos, 2000). Research over the years has

    provided evidence against the healing of tissue (Ghadially et al., 1977) or progressive

    degeneration (Grande et al., 1989) in the case of partial thickness defects. Not only is

    regeneration limited at the cellular level of cartilage, but also negative charges amongst

    proteoglycans during injury contribute to the lack of cellular adhesion (Hendrich et al.,

    2003).

    A blunt trauma to cartilage causes matrix disruption injury. This type of injury may occur

    from a sporting activity or accident. If the extent of injury is not extreme the damage is

    Unive

    rsity

    of Ma

    laya

  • 14

    reversed by the viable chondrocytes present at the site of injury (Temenoff & Mikos,

    2000).

    Thus, despite it being such a serious and significant issue, treatment of cartilage problems

    is severely restricted due to the limited ability of cartilage to self-repair (Lam et al., 2014).

    This often means that degeneration occurs at a higher rate than repair, causing increasing

    failure of the degenerative cartilage surface (O'Driscoll, 1998).

    2.5 Treatment Options for Cartilage Injury

    To provide a sustainable solution to the problem of cartilage damage the primary

    objective of every clinician is to yield normal function of the joint(Temenoff & Mikos,

    2000). Generally there exists three major interventions to restore joint functionality.

    2.5.1 Marrow Stimulation Technique

    The marrow stimulation technique involves micro-fracture, abrasion arthroplasty, or

    drilling, and is usually adopted as a primary treatment for cartilage defects up to 2 to 4

    cm2. This technique involves creating defects that extend to the sub-chondral bone that

    eventually result in clotting at the site of defect and a natural repair response being

    induced. This response leads to the formation of fibrocartilage repair tissue.

    Fibrocartilage tissue is undesirable as it is less resilient than articular cartilage, and

    mechanically speaking the properties fail to match that of the native cartilage (Buckwalter

    & Mankin, 1998a; Caplan et al., 1997). Such marrow stimulation techniques are

    considered to be effective only in younger patients without any prior surgical intervention

    (Demange et al., 2014; Mithoefer et al., 2009), and the size of the defect that can be treated

    by marrow stimulation technique is limited to 2 to 4 cm2. Further, this technique is

    ineffective for the femoral condyle region (Gudas et al., 2005; Kreuz et al., 2006).

    Unive

    rsity

    of Ma

    laya

  • 15

    2.5.2 Osteochondral Grafting

    Tissue transplantation of periosteum, perichondrium or osteochondral grafts is used for

    regeneration of hyaline cartilage (Bouwmeester et al., 1997; Buckwalter & Mankin,

    1998a; Hunziker, 2002). The most common approach is to harvest osteochondral

    cylinders from the donor site and then fit them into holes already drilled in areas of defect.

    The drilled areas are filled via one or more grafts that range from 5 to 11 mm in diameter

    while their lengths are 13 to 15 mm (Hangody, 1997). Studies have shown mixed results

    in cases of osteochondral transplants. Reduced pain and improved function was reported

    after 6.5 years by Outerbridge et al. (Outerbridge et al., 1995). In two separate studies by

    Jakob et al. (Jakob et al., 2002), and Hangody et al. (Hangody et al., 2001) improved knee

    function and good results were reported in more than 90% of patients. In contrast, Laprell

    and Petersen reported a normal knee in fewer than 50% of patients after a follow up of 6

    to 12 years (Laprell & Petersen, 2001). A major limitation of this approach is the

    availability of tissue where there are large cartilage defects. Further, long term follow up

    is missing and this technique is reported to be most appropriate for patients less than 35

    years of age (Buckwalter & Mankin, 1998a; Caplan et al., 1997; Hunziker, 2002; Insall,

    2001). In addition, osteochondral transplantation is associated with some post-operative

    complications. The accumulation of water (joint effusion), bleeding in joint spaces

    (hemarthrosis), and persistent swelling have all been reported after osteochondral

    transplantation (Bobic, 1999; Bös et al., 2000; Hangody et al., 2001; Hunziker, 2002).

    Researchers have also reported loose bodies, donor site pain, and avascular necrosis as

    other post-operative complications that emanate due to osteochondral transplantation

    (Aigner & Fan, 2003).

    Unive

    rsity

    of Ma

    laya

  • 16

    2.5.3 Autologous Chondrocyte Transplantation

    Due to the failure of osteochondral transplantation, autologous chondrocyte

    transplantation was developed and used since 1994 by Brittberg and coworkers (Brittberg

    et al., 1994). Autologous chondrocyte implantation (ACI) is considered to be used as the

    last line of defence when other techniques fail. ACI is considered able to repair full-

    thickness lesions in cartilage up to 16 cm2. This technique involves isolation of a patient’s

    own cells grown in-vitro then implanted at the site of the defect. The follow up results of

    ACI after 2 to 9 years have reported a failure in more than 30% patients after treatment

    of multiple lesions. Furthermore, patients with patella lesion treatment demonstrated

    more than 30% failure, but isolated cartilage lesion treatment displayed less than 10%

    failure (Peterson et al., 2000). Another long-term follow up study by Harris J. D. et al.

    sought to determine if current literature supports ACI over other interventions. They

    observed that ACI can only be helpful over other interventions if the size of defect is

    greater than 4 cm2. ACI or micro fracture or osteochondral transplants, in summary, only

    provide short term success.(Harris et al., 2010) A study by Niemeyer P and co-workers

    reported that complications like graft failure, delamination, chondromalacia (softening of

    cartilage) and hypertrophy can arise after ACI (Niemeyer et al., 2008). More recent

    reports by Harris J. D et al. have reported decreased failure rates (up to 7.5 %) for ACI,

    however there are other reports that continue to demonstrate inconsistent outcomes of

    ACI without any clear recommendation. Furthermore, it was also reported that the

    differences between various treatments were so small that their clinical significance is

    questionable, beyond mere statistical significance (Vavken & Samartzis, 2010). Lately it

    was reported that ACI improves movement significantly after at least 24 months (von

    Keudell et al., 2016).

    Unive

    rsity

    of Ma

    laya

  • 17

    2.6 Cartilage Tissue Engineering

    Current treatment options, like ACI, Osteochondral transplants and marrow stimulation

    have demonstrated variable success rates, however more long-term results are not

    satisfactory (Kreuz et al., 2006; Niemeyer et al., 2008; Redman et al., 2005; Vavken &

    Samartzis, 2010). The major limitation that arises due to the use of the aforementioned

    therapeutic techniques for cartilage repair is the lack of mechanical properties of newly

    formed tissue compared to native articular cartilage tissue. Due to these inferior

    mechanical properties, the newly formed tissue is prone to failure (Hunziker, 2009).

    Cartilage tissue engineering strategies contribute to a durable and functional replacement

    option, beyond any other surgical intervention or technique (Kock et al., 2012).

    Tissue engineering is a 3 tier based approach, involving the combination of living cells,

    a cell signalling strategy, and a scaffold (Hendrich et al., 2003). Cartilage tissue

    engineering allows researchers a great deal of flexibility in combining various techniques

    independently and in combination to address the issue of cartilage repair. Cartilage tissue

    engineering also allows the selection of various cell types to be implemented in-vitro for

    a better understanding of cartilage repair. Moreover, researchers have shown positive

    effect of signalling molecules in enhancing anabolic effects and increased ECM synthesis

    (Elder & Athanasiou, 2009; Seifarth et al., 2009). Further, a widely researched area for

    better fabrication of cartilage tissue is external mechanical stimuli. Direct dynamic

    compression has been demonstrated to increase ECM production while improving the

    compressive properties of the engineered tissue (Bian et al., 2010; Chen et al., 2015; Luo

    et al., 2015). Another leverage given by a cartilage tissue engineering strategy to the

    researchers is modification of the properties of scaffolds. Scaffolds provide a temporary

    frame for the cells to attach to and proliferate while the tissue is being formed and the

    scaffold is being degraded. Recent publications have shown enhanced mechanical and

    biological properties of newly fabricated tissue using polymeric scaffolds (Camarero-

    Unive

    rsity

    of Ma

    laya

  • 18

    Espinosa et al., 2016; Schulze-Tanzil, 2015). Various cartilage tissue engineering studies

    have been undertaken to engineer a tissue with native sGAG content. However, they have

    failed to achieve the collagen content present in native articular cartilage, and which gives

    the cartilage its tensile properties (Eyrich et al., 2007; Kock et al., 2012). Though there

    are many advantages to cartilage tissue engineering, nevertheless finding optimal cell

    source is the first issue, followed by choice of scaffold material, the role and the effect of

    biochemical and mechanical stimulus and lastly tackling any infection that might occur

    due to surgical intervention, amongst others are pitfalls that need to be addressed

    (Bhattacharjee et al., 2015; Kock et al., 2012). Gaut C. and Sugaya K. recently suggested

    that in order to obtain a clinically effective solution for cartilage tissue engineering; a

    combination of factors is required, such as; cell signalling, scaffold material, mechanical

    and biological stimulus must be considered (Gaut & Sugaya, 2015).

    Given the clear advantages of Cartilage Tissue Engineering, but yet the continued

    inability to fully restore cartilage functionality in the long term, the present study seeks

    to further advance this technique toward an effective and lasting repair.

    2.6.1 Cell Sources

    A source of cells is one of the key components of a Tissue Engineering strategy. There is

    no current consensus on the optimal source of cell for cartilage tissue engineering, with

    division between the use of stem cells, fibroblasts, or chondrocytes – each with their

    relative merits and disadvantages.

    Stem cells are particularly attractive due to their differentiation potential and the fact they

    may be harvested from various tissues. Though stem cells overcome the problem of

    limited supply of cell as they can be easily harvested from the patient’s body itself (from

    bone marrow (Boeuf & Richter, 2010) and adipose tissue (Gir et al., 2012)), bone marrow

    stem cells have lower mechanical properties and matrix synthesis when compared to

    chondrocyte seeded scaffolds (Thorpe et al., 2010) and adipose stem cells demonstrate a

    Unive

    rsity

    of Ma

    laya

  • 19

    lesser potential than bone marrow stem cells despite being capable of being differentiated

    into chondrocytes (Kock et al., 2012). Furthermore, embryonic stem cells tend to

    differentiate hypertrophically and moreover, there exists many ethical and legal issues to

    overcome before considering embryonic stem cell use (Jukes et al., 2008). Mesenchymal

    stem cells are another potential source, however they have demonstrated high cartilage

    hypertrophy markers such as Collagen type X (COL X) & Matrix (MMP-13 (Mueller &

    Tuan, 2008).

    Chondrocytes are well suited to cartilage tissue engineering because they are the native

    cells of cartilage tissue (Nicoll et al., 2001). However, they have limited availability and

    generally need to be harvested from a monolayer to get an high number of cells, meaning

    they eventually lose their cell phenotype (Cournil-Henrionnet et al., 2008).

    Another potential source of cells for cartilage tissue engineering are the articular cartilage

    progenitor cells that are useful in producing the amount required for cell culture but do

    not have the capability to produce cartilage matrix (Zhou et al., 2014). It is reported that

    they may also cause donor site morbidity (Mathur et al., 2012).

    A new approach should be using chondrocytes in an in-vitro culture systems that mimics

    the native environment to overcome the limitation of limited cell numbers and allows

    maintenance of cell phenotype.

    2.6.2 Cell Signalling Strategies

    A key aspect of Tissue Engineering is the integration of a cell signalling strategy

    alongside the cellular and scaffold components. This may include aspects such as

    biochemical cues, facilitation or stimulation of cell-cell interactions, biomechanical

    signalling, or manipulation of the environmental factors involved.

    Human body tissues have various associated stimuli that help regulate normal function of

    the tissue. Whether it is electrical, mechanical, or chemical, body tissues undergo

    different kinds of stimulus; heart tissues undergo electrical stimulation (Cannizzaro et al.,

    Unive

    rsity

    of Ma

    laya

  • 20

    2007; Godier-Furnémont et al., 2015), bone tissues are subjected to tension and

    compression (Nyman et al., 2009; Romanos et al., 2015), and similarly, cartilage tissue

    undergoes compression, hydrostatic pressure, and shear strain (Ryan et al., 2009; Schätti

    et al., 2015). Cartilage tissue also experiences a unique stimulus, which is tension in

    oxygen (Portron et al., 2015). This is mainly due to the avascular nature of cartilage tissue

    (Hansen et al., 2001). When considering cartilage tissue, in particular, successful tissue

    engineering will be incomplete without a mechanical stimulus and low oxygen tension.

    2.6.2.1 Mechanical stimulus

    In the last decade, researchers have increasingly realised the importance of external

    mechanical stimuli to enhance the quality of produced cartilage (Temenoff & Mikos,

    2000). However, much conflicting information has been reported regarding how

    chondrocytes behave in response to mechanical stimulation (Lee et al., 2005).

    Physiologically, articular cartilage primarily undergoes compression at joint level (Lee et

    al., 2005). In an average human pressure of up to 1MPa during standing, 0.4MPa while

    walking, and up to 20MPa while standing up from a chair have been reported (Gooch &

    Tennant, 1997; Lee et al., 2005; Urban, 1994). A range of factors are affected by the

    compressive loading of articular cartilage. These include biochemical and physical

    gradients of nutrients, ion concentrations, electrical charge, and pH (Gray et al., 1988;

    Guilak et al., 1995). Different studies have shown that a mechanical stimulus positively

    influences cartilage development (Heath & Magari, 1996). The way ECM is composed

    and organised provides the cartilage with its biomechanical properties. The presence of

    type II collagen helps strengthen the cartilage while proteoglycans build up compressive

    resistance in the cartilage (McMahon et al., 2008). Grodzinsky A.J. and co-workers

    reported that mechanical stimuli on chondrocytes is essential in order to maintain the

    integrity of cartilage (Grodzinsky et al., 2000).

    Unive

    rsity

    of Ma

    laya

  • 21

    In its native environment cartilage is usually compressed to about 10% (Sanchez-Adams

    et al., 2014). Various studies showcase the importance of dynamic compression.

    Researchers have studied the effects of compressing the cartilage from about 5% to 50%

    with frequencies ranging from 0.001 Hz to 1Hz using various polymers and gel constructs

    (Albro et al., 2013; Appelman et al., 2011; Chen et al., 2015; Chowdhury et al., 2006;

    Chowdhury et al., 2003; Davisson, Twana et al., 2002; Démarteau et al., 2003; Di

    Federico et al., 2014; Hunter et al., 2002; Jeon et al., 2012; Kisiday et al., 2004; Li et al.,

    2013; Morel et al., 2005; Piscoya et al., 2005). The majority of the researchers have

    favoured dynamic compression and have demonstrated a higher GAG content with better

    cell proliferation and increased cartilage matrix specific gene expression of Type II

    collage (COL2A1) and Aggrecan (ACAN) (Appelman et al., 2011; Chen et al., 2015;

    Davisson, Twana et al., 2002; Jeon et al., 2012; Kisiday et al., 2004). However,

    contradicting results are present for the amount of compression. Some studies support

    dynamic compression in the range of 0.001% to 31% (Appelman et al., 2011; Chen et al.,

    2015; Davisson, Twana et al., 2002; Démarteau et al., 2003; Jeon et al., 2012) while others

    have reported detrimental effect of dynamic compression ranging from 15% and up to

    50% (Davisson, Twana et al., 2002; Di Federico et al., 2014; Hunter et al., 2002; Li et al.,

    2013; Piscoya et al., 2005). From the current literature it is evident that a dynamic

    compression beyond physiologic levels of 10% has proved to be detrimental for cartilage.

    One thing is noteworthy here: most of the studies executed to study the behaviour of

    cartilage tissue or cartilage cells are performed on agarose, hydrogels, alginate and type I

    collagen gels. All the aforementioned constructs/ gels can only be used in laboratory

    settings and lack the physical capabilities to be implanted clinically. Limited groups have

    reported dynamic compression on polymer/ polymer composite scaffolds which they

    deem to be physically compatible. Another important factor that seems neglected in the

    current study is the physiologic oxygen level. The majority of the studies of dynamic

    Unive

    rsity

    of Ma

    laya

  • 22

    compression in the domain of cartilage tissue engineering were carried out under normal

    oxygen settings without comparing the effect of atmospheric and physiologic oxygen

    concentration on dynamic compression.

    2.6.2.2 Oxygen Tension/Hypoxia

    Research suggests that the nature of articular cartilage is avascular; hence it receives its

    required nutrients and oxygen by synovial fluid following a passive diffusion mechanism

    (Malda et al., 2003). In relation to rest of the body tissues, articular cartilage experiences

    oxygen concentrations in the range of 1% to 10% as compared to 21% for other tissues

    (Co et al., 2014). Chondrocytes are reported to thrive in oxygen concentration range of

    5% to 10% (Grimshaw & Mason, 2000), while losing their activity in anoxic conditions

    because 1% of Oxygen concentration is reported to exist in an abnormal or due to any

    pathological condition (Grimshaw & Mason, 2000; Zhou et al., 2004). Hypoxia Inducible

    Factors are responsible for chondrocytes adapting to low oxygen tension (Fedele et al.,

    2002). Various researchers have reported that hypoxia in chondrocytes has resulted in

    increased synthesis of ECM proteins in in-vitro conditions (Domm et al., 2002; Madeira

    et al., 2015; Yodmuang, Marolt, et al., 2015). Inhibition of collagen type X had also been

    demonstrated by inducing hypoxic conditions. The Collagen type X is major marker of

    hypertrophy in chondrocytes during the chondrogenesis of adipose-derived mesenchymal

    stem cells (Betre et al., 2006; Portron et al., 2015) and of epiphyseal chondrocytes (Chen,

    X. C. et al., 2006). A transcription factor, Hypoxia inducible factor 1 (HIF-1) is mainly

    responsible for regulation of hypoxic response in the cartilage tissue (Schipani et al.,

    2001). HIF-1α is said to be responsible for angiogenesis and glycolysis while at the same

    time causing cells to proliferate (Denko et al., 2003; Goda et al., 2003). Hypoxia further

    plays a role in formation of collagen fibrils with the help of transcription factor HIF-1 α

    (Takahashi et al., 2000). Current research has reported an individual role of hypoxia for

    better engineered cartilage, however, not enough details are present in the current

    Unive

    rsity

    of Ma

    laya

  • 23

    literature as to how hypoxia will behave in combination with other bio-factors for

    cartilage tissue engineering.

    However, many studies have highlighted the importance of hypoxia as a success for tissue

    engineering of cartilage (Meretoja et al., 2013; Munir et al., 2014; Schipani et al., 2001).

    Researchers have shown that hypoxia induces ECM synthesis in in-vitro chondrocyte

    culture (Domm et al., 2002) and also helps chondrocytes to proliferate (Choi et al., 2014).

    Another important research by Schipani E. et al. demonstrated that lack of hypoxia causes

    chondrocytes to terminally differentiate (Schipani et al., 2001). A study by Hansen U. et

    al. reported a combination of hypoxia and hydrostatic pressure to enhance collagen type

    II production while delaying expression of Collagen type I for in-vitro cartilage tissue

    engineering (Hansen et al., 2001). Further research has also displayed the potential of low

    oxygen tension to enhance Aggrecan and Collagen type II gene expression while

    decreasing Matrix Metalloproteinase – 13 (a pro catabolic gene) expression (Parker et al.,

    2013). Most of the studies were executed on chondrocytes on Tri Calcium Phosphate

    (TCP) plates (Hansen et al., 2001), under agarose gel (Chowdhury et al., 2006; Parker et

    al., 2013) or Alginate (Jeon et al., 2012). Others have used PEG (Appelman et al., 2011),

    PGA (Davisson, Twana et al., 2002), or articular cartilage explants (Piscoya et al., 2005).

    However, the above mentioned studies did not used scaffolds able to be used clinically as

    they either lack the requisite mechanical properties or do not mimicked native cartilage

    structure in-vivo.

    2.6.3 Tissue Engineering Scaffolds

    Different natural and synthetic materials have been tested as scaffolds. Scaffolds have

    been developed by natural polymers (Van de Putte & Urist, 1965; Vandeputte & Urist,

    1965), synthetic polymers (Elisseeff et al., 1999; Meinig et al., 1997), composites (Peter

    et al., 1998; Zhang & Ma, 1999) and ceramics (Friedman et al., 1998; Hollinger &

    Battistone, 1986). Synthetic scaffolds have copious benefits such as a greater availability,

    Unive

    rsity

    of Ma

    laya

  • 24

    decreased risk of disease transfer, fabrication as per requirement, and the ability to be

    shaped into various shapes and sizes (Jeuken et al., 2016).

    Just saying that a material should be biocompatible does not solve all the complications.

    A tissue engineered scaffold should be readily available to the surgeon; it should promote

    cell ingrowth as well as it must degrade in correct ratio with formation of new tissue

    (Holmes, 1979; Levine et al., 1997). Also it should be able to fit into irregular places and

    must be soft enough to mould and hard enough to withstand the mechanical loads in daily

    routine activities (Lichte et al., 2011), meaning that the scaffold should possess

    mechanical strengths of site where it needs to be implanted. The architectural properties

    of the scaffold; which includes pore size and porosity, should be in relation to the actual

    tissue (Peter et al., 1998). The scaffold should also promote cell adhesion and

    proliferation (Lichte et al., 2011). Additionally the design of a scaffold should be

    biomimetic, implicating it must imitate all the characteristics of the tissue where it is

    going to be implanted; whether they are physical chemical or biological properties.

    For the aforementioned reasons the polymeric scaffolds are a promising new approach

    towards tissue engineering for repairing cartilage defects. However, use of polymeric

    materials alone or with composites as cartilage tissue engineering scaffolds remains

    largely unexplored.

    Various materials have showed their ability to be used as engineered tissues; this includes

    metals, ceramics, polymers and their combinations (Burg et al., 2000; Hidalgo-Bastida et

    al., 2007; Hollinger & Battistone, 1986; Liu & Ma, 2004). Among these only using

    materials and ceramics independently results in lack of degradability and processability.

    On the contrary, if polymers are used with metals and ceramics they allow tailored

    properties and proper biodegradability. Various scaffolds based on polymers and polymer

    composites are being developed that demonstrate a promising future (Burg et al., 2000;

    Liu & Ma, 2004; Peter et al., 1998).

    Unive

    rsity

    of Ma

    laya

  • 25

    Particularly for cartilage tissue engineering, various polymers and polymer composites

    have been used. Polymers such as Poly (ε-caprolactone) (Li, W.-J. et al., 2005), poly (D,

    L – Lactic acid-co-glycolic acid) [PLGA] and PLGA with Hyaluronic based polymers

    (Yoo et al., 2005), polyurethane based polymers (Grad et al., 2003), poly (ethylene

    glycol)-terephthalate and poly (butylene terephthalate) based polymers (Woodfield et al.,

    2004), poly (D, L – Lactic acid) [PLA] based polymers (Camarero-Espinosa et al., 2016),

    collagen/PLA and Chitosan/PLA polymers (Haaparanta et al., 2014), poly(γ-propargyl-l-

    glutamate) (PPLG) polymers (Ren et al., 2015), Poly (glycerol sebacate) [PGS]

    (Kemppainen & Hollister, 2010), and Polyoctanediol citrate polymers (Jeong et al.,

    2011).

    Among several polymers and polymer composites, Poly 1, 8 Octanediol Citrate (POC)

    stands as a potential candidate due to its mechanical and chemical properties that suffice

    for cartilage tissue engineering applications. POC has proved to enhance cell attachment

    and helps in proliferation of cells, furthermore POC has been shown to enhance matrix

    production when seeded with chondrocytes (Kang et al., 2006). Studies have also reported

    POC polymer to be more effective than other polymers (PGS & polycaprolactone (PCL))

    in enhancing chondrogenesis (Jeong & Hollister, 2010). Moreover, its physical properties

    can be tailored to those of native articular cartilage (Kang et al., 2006; Moutos et al.,

    2007).

    2.7 Poly Octanediol Citrate (POC)

    2.7.1 POC, its structure, synthesis, and form

    Po